• Aucun résultat trouvé

Atherosclerosis severity is not affected by a deficiency in IL-33/ST2 signaling

N/A
N/A
Protected

Academic year: 2022

Partager "Atherosclerosis severity is not affected by a deficiency in IL-33/ST2 signaling"

Copied!
9
0
0

Texte intégral

(1)

Article

Reference

Atherosclerosis severity is not affected by a deficiency in IL-33/ST2 signaling

MARTIN, Praxedis Sina, et al.

Abstract

Interleukin (IL)-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor.

Previous work demonstrated that the systemic administration of recombinant IL-33 reduces the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice by inducing a Th1-to-Th2 shift. The objective of our study was to examine the role of endogenous IL-33 and ST2 in atherosclerosis. ApoE(-/-), IL-33(-/-)ApoE(-/-), and ST2(-/-)ApoE(-/-) mice were fed with a cholesterol-rich diet for 10 weeks. Additionally, a group of ApoE(-/-) mice was injected with a neutralizing anti-ST2 or an isotype control antibody during the period of the cholesterol-rich diet. Atherosclerotic lesion development was measured by Oil Red O staining in the thoracic-abdominal aorta and the aortic sinus. There were no significant differences in the lipid-staining area of IL-33(-/-)ApoE(-/-), ST2(-/-)ApoE(-/-), or anti-ST2 antibody-treated ApoE(-/-) mice, compared to ApoE(-/-) controls. The absence of IL-33 signaling had no major and consistent impact on the Th1/Th2 cytokine responses in the supernatant of in vitro-stimulated lymph node cells. In [...]

MARTIN, Praxedis Sina, et al . Atherosclerosis severity is not affected by a deficiency in IL-33/ST2 signaling. Immunity, Inflammation and Disease , 2015, vol. 3, no. 3, p. 239-246

PMID : 26417439 DOI : 10.1002/iid3.62

Available at:

http://archive-ouverte.unige.ch/unige:86875

Disclaimer: layout of this document may differ from the published version.

1 / 1

(2)

Atherosclerosis severity is not affected by a deficiency in IL-33/ST2 signaling

Praxedis Martin1, 2, Gaby Palmer1,2, Emiliana Rodriguez1,2, Estelle Woldt1,2, Isabelle Mean1,2, Richard W. James3, Dirk E. Smith4, Brenda R. Kwak2, 5, & Cem Gabay1, 2

1Division of Rheumatology, University Hospital of Geneva, Geneva, Switzerland

2Department of Pathology and Immunology, School of Medicine, University of Geneva, Geneva, Switzerland

3Division of Diabetes, Endocrinology and Nutrition, Department of Internal Medicine, University Hospital of Geneva, Geneva, Switzerland

4Inammation Research Department, Amgen Inc., Seattle, Washington, USA

5Division of Cardiology, University Hospital of Geneva, Geneva, Switzerland

Keywords

Atherosclerosis, IL-33, ST2, Th1-to-Th2-shift

Correspondence

Cem Gabay, Division of Rheumatology, University Hospital of Geneva, 26 Avenue Beau-Sejour, 1206 Geneva, Switzerland. Tel : þ41 22 382 35 00; fax :þ41 22 382 35 09; E- mail : cem.gabay@hcuge.ch

Funding information

This study was supported by the Swiss Science Foundation grants (310030_134691 to GP, 310030_135221 to RWJ, 310030_143343/1 to BRK, and 10030_152638 to CG), the Rheumasearch Foundation (CG), and the Institute of Arthritis Research (CG).

Received: 20 February 2015; Revised: 26 March 2015; Accepted: 30 March 2015 Final version published online 10 May 2015.

Immunity, Inflammation and Disease 2015; 3(3): 239–246

doi: 10.1002/iid3.62

Abstract

Interleukin (IL)-33 is a cytokine of the IL-1 family, which signals through the ST2 receptor. Previous work demonstrated that the systemic administration of recombinant IL-33 reduces the development of atherosclerosis in apolipoprotein E-deficient (ApoE/) mice by inducing a Th1-to-Th2 shift. The objective of our study was to examine the role of endogenous IL-33 and ST2 in atherosclerosis.

ApoE/, IL-33/ApoE/, and ST2/ApoE/ mice were fed with a cholesterol-rich diet for 10 weeks. Additionally, a group of ApoE/ mice was injected with a neutralizing anti-ST2 or an isotype control antibody during the period of the cholesterol-rich diet. Atherosclerotic lesion development was measured by Oil Red O staining in the thoracic-abdominal aorta and the aortic sinus. There were no significant differences in the lipid-staining area of IL-33/

ApoE/, ST2/ApoE/, or anti-ST2 antibody-treated ApoE/ mice, compared to ApoE/ controls. The absence of IL-33 signaling had no major and consistent impact on the Th1/Th2 cytokine responses in the supernatant of in vitro-stimulated lymph node cells. In summary, deficiency of the endogenously produced IL-33 and its receptor ST2 does not impact the development of atherosclerosis in ApoE-deficient mice.

Introduction

Interleukin (IL)-33 is the most recently discovered member of the IL-1 cytokine family (see [1] for review). As a dual function protein IL-33 displays both intracellular and extracellular effects. The extracellular effect is executed by binding to its receptor T1/ST2 (ST2), a member of the IL-1 receptor family [2]. IL-33 is mainly expressed as a nuclear protein in stromal cells, including specialized fibroblasts, epithelial, and human endothelial cells, but only to a limited extent in mouse endothelial cells [3, 4]. Upon cell damage during trauma or infection, IL-33 can be released from necrotic cells and act as an alarmin to alert the immune

system [3]. ST2 is predominantly expressed on type 2 innate lymphoid cells, activated Th2 lymphocytes, and mast cells [5–7], but its expression has also been demonstrated on basophils, eosinophils, and dendritic cells. IL-33 induces the production of Th2 cytokines, enhances serum immuno- globulin synthesis and is, therefore, associated with Th2- dependent inflammatory diseases [2].

Atherosclerosis is a chronic inflammatory disease charac- terized by the formation of arterial lesions, which consist of infiltrating T cells, macrophages that convert into foam cells, as well as resident smooth muscle and endothelial cells producing cytokines, growth factors, and other pro- inflammatory mediators [8]. Several studies provide

(3)

evidence for a protective or beneficial role of IL-33 in cardiovascular biology [9, 10]. Of note, the administration of recombinant IL-33 exerted a protective effect in the apolipoprotein E-deficient (ApoE-/-) mouse model of atherosclerosis. IL-33 treatment reduced the lesion size in the aortic sinus and induced a Th1-to-Th2 switch by increasing the levels of the Th2 cytokines IL-4, IL-5, and IL- 13 and decreasing the Th1 cytokine IFNg in serum and lymph node cells. Coadministration of a neutralizing anti- IL-5 antibody with IL-33 prevented the reduction in plaque size, indicating that IL-5 mediated the atheroprotective effect of IL-33. The neutralization of endogenous IL-33 by the administration of soluble ST2, which acts as a decoy receptor for IL-33, exacerbated the development of atherosclerotic plaques in the aortic sinus and led to increased IFNglevels in both serum and the supernatant of in vitro-stimulated peripheral lymph node cells [11]. These data suggested that exogenously administrated IL-33 plays a protective role in the development of atherosclerosis.

However, no study addressed the development of athero- sclerosis in atherosclerotic-prone mice deficient in either IL- 33 or ST2. Therefore, we generated IL-33/ApoE/and ST2/ApoE/ mice for a side-by-side analysis and in addition, treated ApoE/mice with a blocking anti-ST2 or the isotype-matched control antibody in order to investigate a potential protective effect of endogenous IL-33 during atherosclerosis associated with a cholesterol-rich diet. The results described herein show that the deficiency of the endogenous IL-33/ST2-axis does not impact the develop- ment of atherosclerosis in ApoE-deficient mice.

Results and Discussion

Deficiency of endogenous IL-33 signaling does not affect the development of atherosclerosis To examine the effect of endogenous IL-33 signaling on the development of atherosclerosis, IL-33/ApoE/, ST2/

ApoE/, and ApoE/ mice were fed a cholesterol-rich (1.25%, no cholate) diet. In addition, a blocking anti-ST2 antibody or an isotype-matched control antibody were used during feeding of the diet in ApoE/mice (ApoE/, anti- ST2 or ApoE/, isotype control). Efficacy of the blocking anti-ST2 antibody was demonstrated previously [12, 13]. At the beginning of the cholesterol-rich diet, body weight was not different between the groups (Supporting Information Table S1), but there was some variation of serum total cholesterol levels between the groups. (Supporting Informa- tion Table S2). After 10 weeks of cholesterol-rich diet, the body weight increased significantly, but without any significant difference between the groups (Supporting Information Table S1). The cholesterol-rich diet induced a significant increase of serum total cholesterol in all groups

with the exception of ST2/ApoE/ mice (Supporting Information Table S2). The cholesterol levels after 10 weeks of diet were significantly higher in IL-33/ApoE/ mice than in ST2/ApoE/mice.

No significant differences in the extent of atherosclerotic lesions were observed in the thoracic-abdominal aorta among all groups (Fig. 1A, C). The lesion area in the aortic sinus in IL-33/ApoE/mice was significantly increased by 66% (P¼0.0058) as compared to ST2/ApoE/mice, but not different from the one in ApoE/ control mice (P¼0.4349; Fig. 1B, D). No difference in lesion area in the aortic sinus was detected after the treatment of ApoE/ mice with the anti-ST2 antibody compared to the injection with an isotype control antibody (P¼0.7255) or to untreated ApoE/ mice (P¼0.1110). The lesion area in the aortic sinus in ApoE/mice treated with the blocking anti-ST2 antibody was significantly increased by 89%

(P¼0.0018) as compared to ST2/ApoE/mice.

Our results differ from those of Miller et al., who showed that systemically administered IL-33 reduced the develop- ment of atherosclerosis in ApoE/ mice and that the administration of soluble ST2, which neutralizes endoge- nous IL-33 by acting as a decoy receptor, exacerbated the development of atherosclerotic plaques in the aortic sinus [11]. The discrepant results might be explained by different experimental conditions, such as the composition and the duration of the diet, as well as the sanitary status of the animal housing, which are all variables with an impact on the development of atherosclerosis [14, 15]. In contrast to Miller et al., who fed a high-fat diet with low cholesterol (21% lard, 0.15% cholesterol) for 12 weeks to mice maintained in a pathogen-free facility, we fed the mice a high-fat diet with high cholesterol (20% fat [40 kcal%], 1.25% cholesterol) for 10 weeks under conventional conditions. We can, thus, speculate that different housing conditions with a different microbiota and/or a prolonged duration of diet might influence the experimental results.

Another important difference was the use by Miller et al.

of systemic administration of IL-33, which is known to induce Th2 responses [2, 16], a situation which may not necessarily reflect the effects of physiological amounts of endogenous IL-33. Of course, the administration of soluble ST2 should be mimicking IL-33 deficiency. However, we cannot exclude that soluble ST2 binds other proteins or exerts other activities in vivo [17].

Unexpectedly, we also observed a different phenotype in ST2/ApoE/ as compared to IL-33/ApoE/ mice.

Indeed, with ST2 being the receptor for IL-33, one would expect similar phenotypes in ST2/ApoE/ and IL-33/

ApoE/ mice. However, we recently published other discrepant findings between IL-33/and ST2/mice in K/

BxN serum-induced arthritis, with ST2/mice showing a reduced incidence and severity of arthritis compared to both

Deficiency in IL-33 signaling does not affect atherosclerosis severity P. Martinet al.

240 © 2015 The Authors.Immunity, Inflammation and DiseasePublished by John Wiley & Sons Ltd.

(4)

WT and IL-33/ mice [18]. Furthermore, discrepant findings in studies using different ST2/lines, in different studies using the same ST2-/-lines, or between IL-33/and ST2/ mice have already been described by others, especially in the context of allergic airway inflammation [19–

22]. In order to explain the discrepant observations between the IL-33/and ST2/mice observed in K/BxN serum- induced arthritis, we investigated a number of potential

confounding variables. We did not detect any significant impact of ST2 gene targeting on the expression and functionality of other IL-1R family members, nor the expression of miRNA in the vicinity of the ST2 locus.

Another possible explanation might lie in differences in genetic background between the IL-33/ApoE/ and ST2/ApoE/mouse lines. While the IL-33/mouse line was originally backcrossed for more than 10 generations into

Figure 1. Development of atherosclerosis in mice decient in IL-33/ST2 signaling. Representative photographs (A, B) and quantitative analysis (C, D) of atherosclerotic lesion areas in thoracic-abdominal aortas (A, C) and aortic sinuses (B, D) of ApoE/(n¼20), IL-33/ApoE/(n¼25-26), ST2/ApoE/

(n¼9), or ApoE-/-mice injected with either a blocking anti-ST2 (n¼9) or the isotype control antibody (n¼10) after 10 weeks of a cholesterol-rich diet.

All samples were stained with Oil Red O (red) for lipids and sections of aortic sinuses were counterstained with hematoxylin (purple). The extent of atherosclerotic lesions was analyzed by computer-assisted image quantication as percentage of lipid deposition within the total surface area of the thoracic-abdominal aortas (C) or as absolute values of lipid deposition in aortic sinuses (D), respectively. Data are shown as the meanSEM, 1-way ANOVA, and Tukey's post-test for pairwise comparison. Scale bar: 100mm.

(5)

the C57BL/6 background, the backcross of the ST2/ mouse line is less complete with remnants of genetic material from the original 129 mouse strain [18]. Of note, ApoE/ mice in a 129 background have smaller lesions in the aortic roots [23] and the aorta [24] than ApoE/mice in a C57BL/6 background. Another possibility could be that ST2 has other ligands than IL-33 or that IL-33 has unique intracellular activities independent of ST2.

In order to investigate the role of ST2 using a different approach, we thus injected ApoE/mice with a neutraliz- ing anti-ST2 or an isotype matched control antibody during the entire period of the cholesterol-rich diet. Atherosclerotic lesions in the aortic sinus of ApoE/mice treated with the neutralizing anti-ST2 antibody did not significantly differ from those of ApoE/mice treated with the isotype control antibody. This observation and the additional finding that serum total cholesterol significantly increased after 10 weeks on a cholesterol-rich diet in all groups but not in ST2/ApoE/mice suggest that results obtained with the ST2/ApoE/mice should be interpreted with caution.

Expression of IL-33 in cells of the adventitia IL-33 expression has been reported in cells expressing a-smooth muscle actin in the adventitia of atherosclerotic aortas [11]. Herein, we confirm the expression of IL-33 in the nucleus of cells in the adventitia of all groups except for IL-33/ApoE/ mice (Fig 2, upper and middle panels).

Less IL-33-expressing cells are detected in the aortic sinus of ApoE/and C57BL/6 mice fed a chow diet (Fig. 2, lower panels), suggesting that IL-33 expression is either increased in resident cells in the adventitia during atherosclerosis and/

or IL-33-expressing cells are recruited to the adventitia.

The deficiency of endogenous IL-33 signaling has no impact on the Th1/Th2 cytokine profile To determine whether the absence of IL-33 or ST2 promotes a Th1 rather than a Th2 immunological profile as described [11], cytokine responses were assessed in in vitro cultivated sinus-draining mediastinal and peripheral

Figure 2. Expression of IL-33 in aortic sinuses. Representative immunohistochemical localization of IL-33 on sections of aortic sinuses of ApoE/ (cholesterol-rich diet or chow diet), IL-33/ApoE/, ST2/ApoE/, or ApoE/mice injected with either a blocking anti-ST2 or the isotype control antibody (cholesterol-rich diet) and C57BL/6 control mice (chow diet). Scale bars: 500mm for the overview, 100mm for the zoom. A, adventitia; M, media; I, Intima; L, lumen.

Deficiency in IL-33 signaling does not affect atherosclerosis severity P. Martinet al.

242 © 2015 The Authors.Immunity, Inflammation and DiseasePublished by John Wiley & Sons Ltd.

(6)

(including axillary and inguinal) lymph node cells. Levels of IFNg were lower in the supernatant of mediastinal (Fig. 3A), but not peripheral (Fig. 3B) lymph node cells from IL-33/ApoE/ mice compared to ApoE/ con- trols, a difference which is not reflected by the lesion areas.

Therefore, the relevance of this difference in the cytokine production remains arguable. Peripheral lymph node cells from ST2/ApoE/ mice secreted significantly higher levels of both the Th1 cytokine IFNgand the Th2 cytokine IL-5 compared to lymph node cells from ApoE/, IL-33/ApoE/, and ApoE/ mice which received the neutralizing anti-ST2 antibody. We examined also the levels of IL-17A, since an increased concentration of IL-17 has been reported in plasma of unstable angina and acute myocardial infarction patients, suggesting a potential role of IL-17 in atherosclerosis [25]. However, IL-33/ST2 axis deficiency was devoid of any impact on IL-17A secretion. Taken together, the deficiency or blockage of IL-33 or ST2 did not cause major and consistent changes concerning the Th1/Th2 profile.

Concluding remarks

The results presented herein do not support a role of endogenously expressed IL-33 and ST2 in atherogenesis in ApoE-deficient mice.

Materials and Methods

Mice

C57BL/6J Apolipoprotein E-deficient (ApoE/;Apoetm1Unc) mice were obtained from Jackson Laboratory and crossed with C57BL/6 IL-33/mice (B6.129Sv-Il33) obtained from Amgen, Inc. (Thousand Oaks, CA) [18] or C57BL/6 ST2-/- mice (Il1rl1tm1Anjm) obtained from the MRC Laboratory of Molecular Biology (Cambridge, UK) [26] in order to generate IL-33/ApoE/ or ST2/ApoE/ lines, respectively.

Genotyping was performed by PCR by following Jackson Laboratory protocol for ApoE/ mice or as previously described for IL-33/[18]. Genotyping for theSt2gene was

Figure 3.The impact of the deciency of endogenous IL-33 signaling on the Th1/Th2 cytokine prole in lymph node cells. Cytokine expression prole of lymph node cells after 10 weeks of cholesterol-rich diet. Mediastinal (A) and peripheral (B) lymph node cells from ApoE/(n¼14/n¼19–20), IL-33/

ApoE/(n¼20-23/n¼2326), ST2/ApoE/(n¼3/n¼89), or ApoE-/-mice injected with either a blocking anti-ST2 (n¼79/n¼89) or the isotype control antibody (n¼910/n¼910) were stimulated with anti-CD3 and anti-CD28 for 72 h. IFNg, IL-5, and IL-17A protein levels were measured by ELISA in the supernatant. Data are shown as the meanSEM, 1-way ANOVA, and Tukey's post-test for pairwise comparison.

(7)

performed by a 3-primer PCR combining a common reverse primer (5’-GGAAATGCAACCAGAAGTGCACAGG-3’) with forward primers specific for the wild type (5’- GCTGGATAAAGCTATATCATGG-3’) or the KO (5’- GATTGCACGCAGGTTCTC-3’) alleles. All mice were main- tained under conventional conditions in the animal facility of the Geneva University School of Medicine, and water and food were provided ad libitum. Animal studies were approved by the Animal Ethics Committee of the Geneva Veterinarian Office (licence number: 1067/3620/1) and were performed according to the appropriate codes of practice.

Biological reagents

The monoclonal murinized IgG1 blocking anti-ST2 and the isotype matched control antibodies were generated at Amgen, Inc. Efficacy of the blocking anti-ST2 antibody was demonstrated previously [12, 13]. Cell culture media were obtained from Invitrogen Life Technologies (Basel, Switzerland).

Experimental design

Male ApoE/, IL-33/ApoE/, and ST2/ApoE/ mice were used at 10 weeks of age and then placed on a cholesterol-rich diet (20% fat [40 kcal%], 1.25% cholesterol, no cholate; Research Diets, New Brunswick, NJ) for 10 weeks.

Male ApoE/and C57BL/6 mice fed a chow diet for a total of 20 weeks served as controls. In a separate experiment, male ApoE/mice were randomly grouped at 10 weeks of age and injected twice per week intraperitoneally with PBS, 200mg anti-ST2, or 200mg isotype control antibody for 10 weeks during the feeding of the cholesterol-rich diet. As controls, male IL-33/ApoE/mice were injected intra- peritoneally twice per week for 10 weeks with PBS. Mice were weighed before and after diet. Peripheral blood was collected before and after diet. At 20 weeks of age aortas were separated into two parts, of which the thoracic-abdominal parts were fixed in 2% paraformaldehyde and the aortic sinuses were snap-frozen in OCT compound. In order to compare the data obtained in the two independent experi- ments, the values of the lesion areas in the thoracic- abdominal aorta and the aortic sinus of not treated and PBS-injected ApoE/ mice from the first and second experiment, respectively, were compared by unpaired two- tailed Student’s t-test. Since there was no significant difference, the two experiments were pooled.

Oil Red O staining and atherosclerotic lesion analysis

The extent of atherosclerosis was assessed in thoracic- abdominal aortas and aortic sinus cryosections (7mm) with

Oil Red O staining. The fixed thoracic-abdominal aortas were stained with Oil Red O solution (58% isopropanol, 0.2% Oil Red O; Sigma–Aldrich, Buchs, Switzerland) overnight at 48C and opened longitudinally to the iliac bifurcation. The percentage of lipid deposition (red staining) in the thoracic-abdominal aortas was calculated within the total surface area using the Definiens Developer XD Software. Frozen aortic sinus sections were dried at RT, fixed in 10% formalin, rinsed with distilled H2O, and then with 60% isopropanol. The sections were incubated with Oil Red O solution (60% isopropanol, 0.3% Oil Red O) for 15 min at RT, rinsed with 60% isopropanol, and counter- stained with hematoxylin. Slides were scanned with Mirax Scan (Carl Zeiss, Jena, Germany). For the quantification of atherosclerotic lesions in the sinuses, the average of the lesion area from 5 sections distant by 35mm from each other was calculated by measuring the absolute area of lipid deposition using Definiens Developer XD Software. Pictures were taken using the Pannoramic Viewer software (3D HISTECH, Budapest, Hungary).

Serum analysis

Mouse serum total cholesterol concentrations were mea- sured as described [27].

Immunohistochemistry

IL-33 expression was examined on frozen aortic sinus sections (7mm). In brief, after drying at 378C, rehydration in PBS and fixation in 4% paraformaldehyde, endogenous peroxidase activity was blocked and the sections were incubated with a polyclonal goat anti-mouse IL-33 antibody (R&D Systems, Vienna, Austria; at 1mg/mL) as de- scribed [18] and counterstained with hematoxylin. Slides were scanned with Mirax Scan (Carl Zeiss) and pictures taken using the Pannoramic Viewer software (3D HISTECH).

Culture of lymph node cells

Lymph node cells were removed and 1106cells/mL were activated with plate-bound anti-mouse CD3 and anti-mouse CD28 (both 2mg/mL; BD Pharmingen, Allschwil, Switzerland) in RPMI, 10% FCS, 50mMb-ME, 100 U/mL penicillin, and 100mg/mL streptomycin for 72 h.

ELISA

IFN-g, IL-5, and IL-17A protein levels in lymph node cell supernatants were determined using ELISA Ready Set Go kits (IFN-g, IL-17A) from eBioscience (Vienna, Austria) or a DuoSet ELISA kit (IL-5) from R&D Systems.

Deficiency in IL-33 signaling does not affect atherosclerosis severity P. Martinet al.

244 © 2015 The Authors.Immunity, Inflammation and DiseasePublished by John Wiley & Sons Ltd.

(8)

Statistical analysis

Significant variations were calculated using the unpaired or paired two-tailed Student’s t-test or 1-way ANOVA with Tukey’s post-test for pairwise comparison when the ANOVA was statistically significant, as indicated in the figure legends.

P<0.05 was considered significant. Results are expressed as the meanSEM.

Acknowledgments

This study was supported by the Swiss Science Foundation grants (310030_134691 to GP, 310030_135221 to RWJ, 310030_143343/1 to BRK, 10030_152638 to CG), the Rheumasearch Foundation (CG) and the Institute of Arthritis Research (CG). The authors thank Bernard Foglia for helping with the preparation of the aortas, Stephane Jemelin for helping with taking pictures of the thoracic- abdominal aortas, and Mario Kreutzfeld for helping with the quantification.

Author Contributions

PM designed the study, performed experiments, analyzed the data, and wrote the manuscript. GP analyzed data and wrote the manuscript. ER and EW performed experiments and analyzed the data. IM performed experiments and analyzed the data. RWJ analyzed data and wrote the manuscript. DES and BRK analyzed the data and wrote the manuscript. CG supervised the project, designed the study, analyzed the data and wrote the manuscript.

Conflict of Interest None declared.

References

1. Palmer, G., and C. Gabay. 2011. Interleukin-33 biology with potential insights into human diseases. Nat Rev Rheumatol 7:321–329.

2. Schmitz, J., A. Owyang, E. Oldham, Y. Song, E. Murphy, T. K.

McClanahan, G. Zurawski, M. Moshrefi, J. Qin, and X. Li, et al. 2005. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity 23:479–490.

3. Moussion, C., N. Ortega, and J. P. Girard. 2008. The IL-1-like cytokine IL-33 is constitutively expressed in the nucleus of endothelial cells and epithelial cells in vivo: a novel ’alarmin’?

PLoS One 3:e3331.

4. Pichery, M., E. Mirey, P. Mercier, E. Lefrancais, A. Dujardin, N.S Ortega, and J.P. Girard. 2012. Endogenous IL-33 is highly expressed in mouse epithelial barrier tissues, lymphoid

organs, brain, embryos, and inflamed tissues: in situ analysis using a novel Il-33-LacZ gene trap reporter strain. J Immunol 188:3488–3495.

5. Mirchandani, A. S., A. G. Besnard, E. Yip, C. Scott, C. C. Bain, V. Cerovic, R. J. Salmond, and F. Y. Liew. 2014. Type 2 innate lymphoid cells drive CD4þTh2 cell responses. J Immunol 192:2442–2448.

6. Moritz, D. R., H. R. Rodewald, J. Gheyselinck, and R.

Klemenz. 1998. The IL-1 receptor-related T1 antigen is expressed on immature and mature mast cells and on fetal blood mast cell progenitors. J Immunol 161:4866–4874.

7. Lohning, M., A. Stroehmann, A. J. Coyle, J. L. Grogan, S. Lin, J. C. Gutierrez-Ramos, D. Levinson, A. Radbruch, and T.

Kamradt. 1998. T1/ST2 is preferentially expressed on murine Th2 cells, independent of interleukin 4, interleukin 5, and interleukin 10, and important for Th2 effector function. Proc Natl Acad Sci USA 95:6930–6935.

8. Yla-Herttuala, S., J. F. Bentzon, M. Daemen, E. Falk, H. M.

Garcia-Garcia, J. Herrmann, I. Hoefer, S. Jauhiainen, J. W.

Jukema, and R. Krams, et al. 2013. Stabilization of atherosclerotic plaques: an update. Eur Heart J 34:3251–3258.

9. Miller, A. M., D. L. Asquith, A. J. Hueber, L. A. Anderson, W. M. Holmes, A. N. McKenzie, D. Xu, N. Sattar, I. B.

McInnes, and F. Y. Liew. 2010. Interleukin-33 induces protective effects in adipose tissue inflammation during obesity in mice. Circ Res 107:650–658.

10. Hasan, A., F. Al-Ghimlas, S. Warsame, A. Al-Hubail, R.

Ahmad, A. Bennakhi, M. Al-Arouj, K. Behbehani, M. Dehbi, and S. Dermime. 2014. IL-33 is negatively associated with the BMI and confers a protective lipid/metabolic profile in non- diabetic but not diabetic subjects. BMC Immunol 15:19.

11. Miller, A. M., D. Xu, D. L. Asquith, L. Denby, Y. Li, N. Sattar, A. H. Baker, I. B. McInnes, and F. Y. Liew. 2008. IL-33 reduces the development of atherosclerosis. J Exp Med 205:339–346.

12. Ngoi, S. M., M. C. St Rose, A. M. Menoret, D. E. Smith, M. G.

Tovey, A. J. Adler, and A. T. Vella. 2012. Presensitizing with a Toll-like receptor 3 ligand impairs CD8 T-cell effector differentiation and IL-33 responsiveness. Proc Natl Acad Sci USA 109:10486–10491.

13. Beamer, C. A., T. A. Girtsman, B.P. Seaver, K. J. Finsaas, C. T.

Migliaccio, V.K. Perry, J.B. Rottman, D. E. Smith, and A.

Holian. 2013. IL-33 mediates multi-walled carbon nanotube (MWCNT)-induced airway hyper-reactivity via the mobili- zation of innate helper cells in the lung. Nanotoxicology 7:1070–1081.

14. Danzaki, K., Y. Matsui, M. Ikesue, D. Ohta, K. Ito, M.

Kanayama, D. Kurotaki, J. Morimoto, Y. Iwakura, and H.

Yagita, et al. 2012. Interleukin-17A deficiency accelerates unstable atherosclerotic plaque formation in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol 32:273–280.

15. Stepankova, R., Z. Tonar, J. Bartova, L. Nedorost, P.

Rossman, R. Poledne, M. Schwarzer, and H. Tlaskalova- Hogenova. 2010. Absence of microbiota (germ-free

(9)

conditions) accelerates the atherosclerosis in ApoE-deficient mice fed standard low cholesterol diet. J Atheroscler Thromb 17:796–804.

16. Kondo, Y., T. Yoshimoto, K. Yasuda, S. Futatsugi-Yumikura, M. Morimoto, N. Hayashi, T. Hoshino, J. Fujimoto, and K Nakanishi. 2008. Administration of IL-33 induces airway hyperresponsiveness and goblet cell hyperplasia in the lungs in the absence of adaptive immune system. Int Immunol 20:791–800.

17. Sweet, M. J., B. P. Leung, D. Kang, M. Sogaard, K. Schulz, V.

Trajkovic, C. C. Campbell, D. Xu, and F. Y. Liew. 2001. A novel pathway regulating lipopolysaccharide-induced shock by ST2/T1 via inhibition of Toll-like receptor 4 expression.

J Immunol 166:6633–6639.

18. Martin, P., D. Talabot-Ayer, C. A. Seemayer, S. Vigne, C.

Lamacchia, E. Rodriguez, A. Finckh, D. E. Smith, C. Gabay, and G. Palmer. 2013. Disease severity in K/BxN serum transfer-induced arthritis is not affected by IL-33 deficiency.

Arthritis Res Ther 15:R13.

19. Hoshino, K., S. Kashiwamura, K. Kuribayashi, T. Kodama, T.

Tsujimura, K. Nakanishi, T. Matsuyama, K. Takeda, and S.

Akira. 1999. The absence of interleukin 1 receptor-related T1/ST2 does not affect T helper cell type 2 development and its effector function. J Exp Med 190:1541–1548.

20. Coyle, A. J., C. Lloyd, J. Tian, T. Nguyen, C. Erikkson, L.

Wang, P. Ottoson, P. Persson, T. Delaney, and S. Lehar, et al.

1999. Crucial role of the interleukin 1 receptor family member T1/ST2 in T helper cell type 2-mediated lung mucosal immune responses. J Exp Med 190:895–902.

21. Mangan, N. E., A. Dasvarma, A. N. McKenzie, and P. G.

Fallon. 2007. T1/ST2 expression on Th2 cells negatively regulates allergic pulmonary inflammation. Eur J Immunol 37:1302–1312.

22. Oboki, K., T. Ohno, N. Kajiwara, K. Arae, H. Morita, A. Ishii, A. Nambu, T. Abe, H. Kiyonari, and K. Matsumoto, et al.

2010. IL-33 is a crucial amplifier of innate rather than acquired immunity. Proc Natl Acad Sci USA 107:18581–

18586.

23. Maeda, N., L. Johnson, S. Kim, J. Hagaman, M. Friedman, and R. Reddick. 2007. Anatomical differences and athero- sclerosis in apolipoprotein E-deficient mice with 129/SvEv and C57BL/6 genetic backgrounds. Atherosclerosis 195:

75–82.

24. Paigen, B., A. Morrow, C. Brandon, D. Mitchell, and P.

Holmes. 1985. Variation in susceptibility to atherosclerosis among inbred strains of mice. Atherosclerosis 57:65–73.

25. Hashmi, S., and Q. T. Zeng. 2006. Role of interleukin-17 and interleukin-17-induced cytokines interleukin-6 and interleu- kin-8 in unstable coronary artery disease. Coron Artery Dis 17:699–706.

26. Townsend, M. J., P. G. Fallon, D. J. Matthews, H. E. Jolin, and A. N. McKenzie. 2000. T1/ST2-deficient mice demonstrate the importance of T1/ST2 in developing primary T helper cell type 2 responses. J Exp Med 191:1069–1076.

27. James, R. W., and D. Pometta. 1990. Differences in lipoprotein subfraction composition and distribution be- tween type I diabetic men and control subjects. Diabetes 39:1158–1164.

SUPPORTING INFORMATION

Additional supporting information may be found in the online version of this article at the publisher’s web-site.

Table S1: Body weight (g) before and after cholesterol-rich diet.

Table S2: Serum total cholesterol concentrations (mM) before and after cholesterol-rich diet.

Deficiency in IL-33 signaling does not affect atherosclerosis severity P. Martinet al.

246 © 2015 The Authors.Immunity, Inflammation and DiseasePublished by John Wiley & Sons Ltd.

Références

Documents relatifs

We studied the forms of Aβ and their interaction with the Apoe fragments in post-mortem brains from control and AD patients by western blots and co-immunoprecipitation..

From their analysis as a whole, the authors concluded that ‘‘differences in mycorrhizal responsiveness measured among inbred lines are deter- mined primarily by variation

investigate the extent of the ε4 advantage in young adults across a broad coverage of cognitive domains, and also, using a double-blind placebo-controlled study, to explore

For the first set of analyses, we used the extracted FDG-PET values from the 4 areas showing a significant APOE e4 3 years of education interaction so as to (1) assess

A systematic press articles’ analysis from 1977 to 2006 shows that the rising number of these collective actions since the early 1990s may have gradually increased the

Whereas rIL-33 treatment was associated with an early increase of parasite loads (Mann-Whitney test: U = 5, P &lt; 0.05 on D15) in BALB/c, they decreased until Day 60, where they

20 Autoradiography of 64 Cu-labelled nanostructured lipid carriers (NLC) and Oil Red O histology (neutral lipids staining) of aortas showed significant particle uptake 21

Conclusion: Our results showed that (i) the APOE epsilon polymorphism has the expected impact on the plasma lipid profile and (ii) the rs4420638 G allele may counterbalance